- Research
- Open access
- Published:
Acute vascular and cardiac effects of lenvatinib in mice
Cardio-Oncology volume 11, Article number: 14 (2025)
Abstract
Background
Tyrosine kinase inhibitors (TKIs) targeting vascular endothelial growth factor (VEGF) receptor signalling are used in cancer therapy to inhibit angiogenesis. Unfortunately, VEGF inhibitors are known to induce severe hypertension in patients. This study aimed to elucidate the impact of the TKI lenvatinib on blood pressure, arterial stiffness, vascular reactivity, as well as cardiac function in a short-term murine model to shed light on potential contributors to cardiovascular (CV) toxicities associated with VEGF inhibition.
Methods
Male C57BL/6J mice were randomly divided into 2 cohorts, either treated for 4 days with lenvatinib 4 mg/kg/day or 40% hydroxypropyl β-cyclodextrin as control. In an additional study, mice were subjected to a 4-day treatment followed by a 4-day wash-out, with echocardiography and blood pressure measurements performed on day 2 and 7. Subsequently, ex vivo vascular reactivity of thoracic aortic segments was determined.
Results
Lenvatinib induced hypertension and arterial stiffness (i.e., increased pulse wave velocity), starting from day 2 of treatment. Further, left ventricular ejection fraction was reduced and the ventricle dilated upon treatment. Lenvatinib induced neither endothelial dysfunction nor impaired vascular smooth muscle cell reactivity to nitric oxide (NO). Interestingly, lenvatinib demonstrated a concentration-dependent increase in ATP-mediated relaxation. In addition, after the 4-day wash-out period, lenvatinib-treated mice did not show complete remission of hypertension. However, arterial stiffness, ATP-mediated relaxation and cardiac adaptation were recovered.
Conclusion
This comprehensive investigation provides valuable insights into the interplay between VEGF inhibition, vascular function and cardiac outcomes, emphasising the need for nuanced understanding and further exploration of the differential effects of lenvatinib on the CV system. Additionally, the study proposes a synergistic formation between VEGF and ATP, indicating an enhanced response via P2Yx receptor signalling.
Introduction
Neo-angiogenesis, mainly relying on vascular endothelial growth factor (VEGF) and its receptor-2 (VEGFR-2), is crucial in supplying primary tumours with blood and nutrients [1]. Overexpression of VEGFR-2 is seen in the majority of human tumours, contributing to high vascular density and metastasis, which correlate with poor cancer prognosis [2, 3]. Targeting tumour angiogenesis by using tyrosine kinase inhibitors (TKIs), and therefore inhibiting VEGFR-2, has improved survival rates in cancer patients [4]. One of the most potent and clinically-used TKIs targeting VEGFR-2 is lenvatinib [5, 6]. While it has been demonstrated to improve clinical outcomes [7], undesirable cardiovascular (CV) toxicities have emerged [4].
The most reported CV adverse effect of VEGFR-2 inhibition is hypertension, which often results in dose reduction or interruption of a potentially lifesaving cancer treatment [4]. VEGFR-2 inhibitor-induced hypertension appears to be mediated by both structural and functional changes in the vascular endothelium [8]. Functional changes were initially hypothesised to be due to reduced endothelium-dependent vasodilation [8]. Normally, VEGF binding to VEGFR-2 activates the intracellular signalling cascade leading to stimulation of the phosphoinositide 3-kinases /protein kinase B pathway, which enhances endothelial nitric oxide synthase (eNOS) phosphorylation and results in nitric oxide (NO) release [9]. TKIs inhibit the interaction between VEGF and its receptor leading to impaired NO production [8, 9]. Additionally, patients treated with VEGFR-2 inhibitors showed increased plasma levels of endothelin-1, which is known to reduce NO production and increase reactive oxygen species production, also contributing to hypertension development [9]. While several studies support the role of endothelial dysfunction and reduced NO production in CV toxicity, there is no consensus on the exact mechanism of VEGFR-2 inhibitors-induced CV complications [8,9,10,11,12,13,14,15].
Additionally, TKIs have been associated with structural remodelling and/or fibrosis, which induces microvascular rarefaction and arterial stiffening [9]. Patients treated with TKIs, such as sunitinib and sorafenib, showed increased arterial stiffness, measured as pulse wave velocity (PWV), shortly after treatment initiation but this phenomenon normalises after discontinuation [16, 17]. The rapid onset and reversibility of vascular stiffness associated with TKIs suggest an acute mechanism rather than structural remodelling as a potential cause of the reduced arterial compliance induced by these drugs, but further studies are needed. Additionally, TKIs have also been linked to CV toxicities [18, 19], although its mechanism(s) remains elusive [20]. Clinically, sorafenib, sunitinib, pazopanib and lenvatinib, were associated with cardiac dysfunction [21].
VEGF inhibition is known to induce hypertension in patients, yet its effect on arterial stiffness and cardiac function remains unclear. Here, we aimed to address this gap by investigating the effect of lenvatinib on both aortic biomechanics and reactivity, as well as on cardiac function. This study would add to the current knowledge on CV complications when using VEGF inhibitors.
Methods
Animals and ethical approval
Male 10-week-old C57BL/6J mice (n = 32) weighing between 25 and 30 g were used in this study. All surgical and experimental procedures were conducted with approval of the Ethical Committee for Animal Testing of the University of Antwerp (ethical file 2022–40) conformed with the ARRIVE guidelines, under Directive 2010/63/EU, and with the Belgian Royal Decree of 2013.
Study design
Mice were administered lenvatinib (4 mg/kg, Sigma-Aldrich, SML3017) via intravenous injection for 4 consecutive days. The control group received vehicle (40% 2-hydroxypropyl-β-cyclodextrin, Sigma-Aldrich, H107, in sterile saline) in the same volume as the lenvatinib group. The dose for lenvatinib was chosen based on previous studies showing a hypertensive effect following intravenous injection.
In a first cohort (Treatment), cardiac function was investigated in vivo on day 2. On day 4, ex vivo vascular reactivity and left ventricular pressure (LVP) were analysed. In a second cohort, CV function was evaluated on day 2 day after treatment, as well as on day 3 of the wash-out period (day 7), followed by ex vivo evaluation of vascular reactivity on day 8. On day 2 and day 4, measurements were performed 5 h after compound or vehicle injection. A schematic overview of the study design is presented in Fig. 1.
Mice treatment plan and experimental procedure. Animals were divided into two cohorts. Both cohorts received daily treatments of either 4 mg/kg lenvatinib or 40% (2-Hydroxypropyl)-β-cyclodextrin (vehicle) for 4 days. Echocardiography was performed on day 2 for both cohorts. Cohort 2 had additional blood pressure measurements on day 2. On day 4, cohort 1 underwent terminal in vivo LVP measurements and ex vivo vascular reactivity assessments. During wash-out, in vivo echocardiography and blood pressure measurements were done on day 7, and ex vivo vascular reactivity was assessed on day 8. Dashed line indicated in vivo measurements; Solid line indicate terminal experiments. HPβCD = 40% (2-Hydroxypropyl)-β-cyclodextrin; LVP = left ventricular pressure
Echocardiography
Echocardiographic evaluation (n = 8 /group/cohort) was performed to assess the impact of lenvatinib on vascular function (PWV), cardiac function (left ventricular function) and structure. Echocardiographic measurements of cohort 1 were performed with a Vevo2100 high-frequency ultrasound system (Visual Sonics), while all measurements of cohort 2 were performed with the Vevo F2 LAZR-X imaging station (Visual Sonics). To perform the measurements, mice were anesthetised using isoflurane (3% for induction and 1–1.5% for maintenance). During the procedure, heart rate, and respiratory rate as well as body temperature, were constantly measured. A detailed description of the method was previously published [22].
Left ventricular pressure analysis
Left ventricular pressure (LVP) analysis was conducted as previously described [22]. Sevoflurane was chosen as anaesthetic agent at 8% for induction and 4–5% for maintenance.
Peripheral blood pressure measurements
Systolic blood pressure, diastolic blood pressure, and mean arterial pressure were non-invasively measured in restrained, awake mice using a tail-cuff system equipped with a programmed electro sphygmomanometer (Coda, Kent Scientific Corporation, Torrington, USA). Before the actual measurements, mice underwent training to minimise stress and variability. During this training, the cuff system was applied to the mouse tail exactly as it would be during the real measurements. Training and measurement sessions lasted 20 min each.
Ex vivo vascular reactivity
Isometric organ bath
Thoracic descending aortic segments were procured and mounted at a preload of 20 mN in the bath. The experimental protocol was started after a 60 min equilibration period to allow optimal stabilisation in Krebs–Ringer solution at 37 °C. Vascular smooth muscle cell (VSMC) contraction was evaluated by adding cumulative concentrations of phenylephrine (PE) (Sigma-Aldrich, Overijse, Belgium), an α1-adrenergic receptor agonist.
Afterwards, endothelium-dependent vasodilation was assessed by addition of cumulative concentrations of acetylcholine (Sigma-Aldrich, Overijse, Belgium) (ACh; 3 nM–10 μM), a muscarinic receptor agonist. Similarly, endothelium-dependent relaxation responses were also assessed by adding cumulative concentrations of adenosine triphosphate (ATP), a purinergic receptor agonist, on pre-contracted segments (with PE 2 µM).
For a final measurement, the segments were stimulated with Nω-nitro-L-arginine methyl ester (Sigma-Aldrich, Overijse, Belgium) (L-NAME; 300 µM), an inhibitor of endothelial nitric oxide synthase (eNOS). After 20 min, cumulative concentrations of PE (3 nM to 10 μM) were given followed by cumulative doses (0.3 nM–10 μM) of the exogenous nitric oxide (NO)-donor diethylamine NONOate (DEANO, Sigma-Aldrich Overijse, Belgium) to evaluate endothelium-independent vasodilation of VSMCs through the cGMP-mediated pathway.
Measuring ex vivo arterial stiffness
The Rodent Oscillatory Set-up to Study Arterial Compliance (ROTSAC) is a custom-built system designed to measure aortic biomechanics and vascular reactivity [23]. It involves mounting 2 mm aortic segments between two wire hooks in 10 mL organ baths. A force-and-length transducer controls and records the force and displacement of the upper hook. The segments oscillate between preloads at 10 Hz, with data sampled at 400 Hz. The vessel's inner diameter is calculated from the inter-hook distance, directly proportional to the inner circumference.
Prior to the experiment, inter-hook distance and vessel length were measured statically at six different preload values (10, 20, 30, 40, 50, and 60 mN) using a camera and calibrated imaging software. The average segment length during each cycle (100 ms) was obtained from the inner diameter-vessel length relationship through linear regression to account for the reduction in vessel length with increasing extrapolated diameter. The transmural pressure was then calculated using the distension force and vessel dimensions.
The Peterson’s elastic modulus (Ep), calculated as a measure of arterial stiffness, was evaluated at different pressures, and performed under physiological conditions (Krebs–Ringer solution), as well as following contraction with α1-adrenergic receptor agonist phenylephrine and relaxation with DEANO.
Thoracic descending aortic segments were subjected to increasing and decreasing pressures under physiological conditions (Krebs–Ringer solution). After loading/unloading pressure steps under physiological condition, aortic segments were then treated with L-NAME (300 μM) to inhibit eNOS and therefore remove the influence of NO. Thereafter, 2 μM PE was added to the organ bath, and the loading/unloading pressure steps were repeated, before treatment with DEANO (2 μΜ) and subsequent alterations of pressure conditions.
Histology
After dissection, cardiac tissue was immediately fixated in 4% formaldehyde (Merck, Overijse, Belgium) for 24 h. Afterwards, the tissues were embedded in paraffin (Leica Biosystems, Diegem, Belgium) and transversely cut into 5 µm sections. Immunohistochemical staining with a primary antibody against laminin (1:1000; lmNB300-144 Novus Biologicals, Abingdon, UK) was used to quantify cardiomyocyte size. The biotinylated secondary antibody was goat anti-rabbit (1:200; Vector Laboratories, Burlingame, USA).
An Olympus BX40 microscope and Universal Graph 6.1 software were used to acquire images, which were quantified using ImageJ software. For each heart, five regions were analysed. Next, 20 cardiomyocytes were randomly selected per region for semi-automated evaluation of the cross-sectional area (final average of 100 measurements).
Vascular mRNA expression (qRT-PCR)
Quantitative real-time polymerase chain reaction (qRT-PCR) was performed to determine vascular mRNA expression of P2Y2 (Mm02619978_s1) and P2Y6 (Mm02620937_s1) purinergic receptors using TaqMan™ probes (Thermo Fisher Scientific) and the TaqMan™ Fast Advanced Master Mix (Applied Biosystems™, Thermo Fisher Scientific). The RNA was extracted from the thoracic aortic segments by using the ISOLATE II RNA Micro Kit (Bioline) in accordance with the manufacturer's instructions and reverse transcribed into cDNA using a cDNA synthesis kit (TaqMan™ Reverse Transcription Reagents, Invitrogen). The reaction was performed using the Quantum studio™ 3 PCR-machine (Applied Biosystems™). Relative mRNA expression (target gene/ β-actin reference gene) of P2Y2 and P2Y6 was reported as fold-change calculated using the ΔΔCT method.
Statistical analysis
All data were analysed using Prism 10.2.2 (GraphPad Software, La Jolla, CA, USA) and expressed as mean ± SEM, with n representing the number of mice. Statistical tests are specified in the figure legends. Statistical significance was defined as *p < 0.05, **p < 0.01, ***p < 0.001 and ****p < 0.0001; ns stands for not significant.
Results
Lenvatinib reversibly increases both blood pressure and arterial stiffness
Systolic and diastolic blood pressure were higher on day 2 in the lenvatinib-treated group (*p < 0.05, Fig. 2A&B). No changes between groups were seen on day 7 (i.e., after the treatment was stopped).
Blood pressure changes in lenvatinib-treated mice on day 2 and 7. Lenvatinib increased both systolic and diastolic blood pressure in mice. A SBP = systolic blood pressure; (B) DBP = diastolic blood pressure; Data are represented as mean ± SEM. N = 8 per cohort. Two-way ANOVA was conducted for between-group comparison (*p < 0.05)
Additionally, abdominal PWV, a marker for arterial stiffness, was increased on day 2 in lenvatinib-treated animals (4.8 ± 0.3 m/s vs. 2.5 ± 0.2 m/s, ****p < 0.0001, Fig. 3A), while no difference was observed after treatment interruption on day 7. Pulse pressure was not altered by the treatment (Fig. 3B). No effects of lenvatinib were observed ex vivo on aortic stiffness in either active or passive state (Fig. 3C&D).
Blood pressure changes and pressure dependency of Peterson’s elastic modulus in lenvatinib-treated mice. Lenvatinib increased both systolic and diastolic blood pressure in mice. (A) PWV = pulse wave velocity; (B) PP = pulse pressure. Pressure dependency of Peterson’s elastic modulus (EP) was measured after stimulation with 2 μM PE in the presence of 300 μM L-NAME (C) or after relaxation with 2 μM DEANO (D). Data are represented as mean ± SEM. N = 7–8 per group. Two-way ANOVA was conducted for between-group comparison (****p < 0.0001)
Lenvatinib increased ATP-dependent vascular relaxation
Vascular reactivity was evaluated ex vivo to investigate whether treatment with lenvatinib alters endothelial function. Lenvatinib did not alter PE-induced isometric contraction (7.8 ± 0.9 mN vs. 7.9 ± 0.8 mN). Even maximum contraction (i.e., PE contraction in the presence of L-NAME) was similar among the treatment groups (Supplementary Fig. 1A-B). Additionally, lenvatinib did not affect ACh-induced endothelium-dependent relaxation (Supplementary Fig. 1C). However, the vasodilator response to ATP was significantly increased after 4 days of lenvatinib treatment (*p < 0.05, Fig. 4A) with a shift in—logIC50 from 4.13 ± 0.32 to 4.82 ± 0.18, yet normalised after wash-out at day 8 (Fig. 4C). DEANO-induced endothelium-independent relaxation was not affected by lenvatinib at any time point (Fig. 4B&D). Vascular mRNA expression of the genes encoding for P2Y6 and P2Y2 receptors was significantly increased in the lenvatinib-treated group on day 2 (***p < 0.001, **p < 0.01, respectively, Fig. 4E&F). On day 8, no effects on the expression of P2Y2 and P2Y6 were observed between the groups.
Relaxation of thoracic aortic segments and aortic mRNA expression of P2Y2 receptor and, P2Y6 receptor in lenvatinib-treated mice on day 4 and 8. Following maximal contraction with PE, VSMC relaxation was assessed in response to increasing concentrations of ATP (A&C), or DEANO (B&D). Lenvatinib increased both receptor mRNA expression on day 2 but not on day 8 (E&F). Vascular mRNA expression is normalised to the internal reference gene β-actin and is expressed relative to the vehicle-treated group. ATP = adenosine triphosphate; DEANO = diethylamine NONOate; dr = dose–response. N = 6 per group. Data are mean ± SEM. Data were analysed using a Two-way ANOVA with Dunnett’s post hoc test was performed (*p < 0.05, **p < 0.01, ***p < 0.001)
Lenvatinib induces cardiotoxicity
Echocardiographic data of mice from cohorts 1 and 2 were performed using two different machines. Therefore, we did not combine the measurements on day two of both cohorts. Systolic ventricular function was reduced at day 2 of treatment, with a drop of 24% in left ventricular ejection fraction (LVEF) in cohort 1 (***p < 0.001, Fig. 5A). Lenvatinib-treated mice of cohort 2 showed a similar reduction in LVEF of 17% (****p < 0.0001, Fig. 5B). On day 7, LVEF was similar in the lenvatinib group compared to control animals. A similar pattern was observed for fractional shortening. Further, diastolic function (E/A and E/E`) was not affected on day 2. At day 7, E/E` was not altered, but mitral valve E/A ratio was increased in lenvatinib-treated mice (Table 1, *p < 0.05).
Echocardiography in lenvatinib-treated mice on day 2 (cohort 1 & 2) and 7 (cohort 2). Lenvatinib caused a reversible reduction in LVEF. A LVEF of mice from cohort 1 was measured on day 2. B LVEF in mice from cohort 2 on both examination days. LVEF = left ventricular ejection fraction. Data is represented as mean ± SEM. N = 8 per cohort. A Mann–Whitney test (B) Two-way ANOVA (***p < 0.001, ****p < 0.0001)
At day 2, left ventricular internal diameter at diastole and systole of lenvatinib-treated mice was significantly larger when compared to control animals (7.3% increase during diastole and 18.6% increase during systole) (*p < 0.05, **p < 0.01, Table 1). While no difference was observed in left ventricular anterior and posterior wall thickness during diastole, systolic wall thickness was significantly reduced in the lenvatinib-treated mice (Table 1), with a reduction of 17.3% during diastole and 22.3% during systole (*p < 0.05, ***p < 0.001, Table 1). During the wash-out phase, the changes in left ventricular dimensions were no longer detected.
Lenvatinib did not alter LVP on day 4
The assessment of lenvatinib-induced effects on hemodynamic parameters via LVP measurements did not show a significant difference between control and treated animals in terms of end-diastolic and end-systolic pressure (Fig. 6A&D). In addition, lenvatinib did not affect ventricular relaxation, as reported by the absence of variations in the time constant of left ventricular isovolumic relaxation (Tau, a measure of diastolic performance) and on the peak rate of pressure increase (dP/dtmin) between the vehicle and the lenvatinib group (Fig. 6B&C). Lenvatinib did not significantly alter the maximum rate of increase in LVP (dP/dtmax).
Indices of diastolic (A, B, C) and systolic function (D, E) derived from LVP analysis in lenvatinib-treated mice. (A) EDP = end-diastolic pressure; (B) dP/dtmin = peak rate of pressure decreases in the ventricle; (C) Tau = isovolumetric relaxation constant; (D) ESP = end-systolic pressure; (E) dP/dtmax = peak rate of pressure rises in the ventricle; Data points are means; vertical bars represent SEM. N = 10 for lenvatinib and N = 10 for control. Unpaired t-test with Welch’s correction was performed for between-group comparison
Lenvatinib treatment increases cardiomyocyte cross-sectional area
Cardiomyocyte cross-sectional area was assessed by staining heart cross-sections for the ubiquitous non-collagenous connective tissue glycoprotein laminin (Fig. 7A-D). Treatment with lenvatinib was associated with a significant increase in the cross-sectional cardiomyocyte area of 62% compared to the control group (***p < 0.001, Fig. 7E).
Laminin immunohistochemical staining of cardiac tissue to quantify cardiomyocyte cross-sectional area. Representative images of cardiomyocyte cross-sectional area following 4-day treatment with vehicle (40% HPβCD) (A, B), and lenvatinib (4 mg/kg) (C, D). Scale bar = 50 µm. E Average of 100 analysed cardiomyocyte cross-sectional area per mouse. CM = cardiomyocyte. Data points are means; error bars represent SEM. Mann–Whitney test was performed (***p < 0.001)
Discussion
This study focused on lenvatinib-induced changes in arterial stiffness, vascular reactivity and blood pressure in mice, in addition to investigating its effect on cardiac function and structure. Lenvatinib elicited a consistent increase in blood pressure and aortic stiffness (PWV) after 2 days of treatment, which was reversible upon discontinuation. Lenvatinib improved ATP-dependent VSMCs relaxation ex vivo in the thoracic aorta. Additionally, lenvatinib reduced LVEF on day 2, which normalised after treatment interruption.
Hypertension
TKIs are known to induce hypertension in (cancer) patients [4]. As such, there is a need for robust preclinical models to investigate TKI-induced hemodynamic alterations. It has been shown that hypertension can be induced by cediranib, sorafenib, pazopanib, vandetanib [24], axitinib, and lenvatinib [15], of which the latter aligns with the findings presented in this study. These findings point to a class effect rather than a unique property of lenvatinib. The rapid onset of hypertension (on day 2 after treatment initiation) suggests an acute vascular response, possibly due to the immediate effect of VEGFR-2 inhibition on endothelial cells (EC) and subsequent reduction in NO availability [8, 13]. However, TKIs also act on VEGF receptors in the renal glomerulus, leading to damage in the glomerular filtration barrier [25, 26] and proteinuria. The renal dysfunction may further contribute to the development of hypertension [9]. Nevertheless, multiple studies suggest that the rise in blood pressure precedes kidney damage and occurs at lower doses of VEGFR inhibitors (reviewed in [9]). Thus, the acute increase in blood pressure observed in this study is likely not affected by renal dysfunction.
In the present study, no effect of lenvatinib on either basal NO or NO-dependent vasorelaxation was observed (2 days after the hypertension measurement). It is important to note that while we observed no changes in NO-related vasoreactivity, our ex vivo experimental conditions may suffer from a washout of lenvatinib in the organ bath, potentially masking relevant NO-related effects.
Arterial stiffening
Hypertension can result from vascular remodelling leading to increased arterial stiffness. Notably, arterial stiffness is a strong and independent predictor of CV morbidity and mortality [27, 28]. Increased PWV, suggestive of increased arterial stiffness, was observed in sorafenib-treated patients within 4 weeks of treatment and was still present after 10 months of therapy [16]. Similarly, sunitinib has been associated with hypertension and increased arterial stiffness after 3.5 weeks of treatment in patients with metastatic renal cell carcinoma [17]. These clinical data suggest that vascular stiffness may contribute to or is the result of VEGFR-2-induced hypertension, although the relationship between the rise in blood pressure and vascular remodelling remains controversial [16]. Indeed, arterial stiffness can be both a cause and a consequence of hypertension with these two events showing a bidirectional relationship [29]. Mechanisms of arterial stiffness include not only chronic events, such as fibrosis, reduced elastin-collagen ratio, extracellular matrix calcification, and VSMC phenotype switching [30], but also acute changes in VSMC contractility or increased EC-Matrix adhesion [9, 31, 32]. We did not observe differences in aortic contraction induced by PE between the groups. Although the pathophysiological events underlying VEGFR-2-induced arterial stiffness have not been investigated, the rapid onset of vascular stiffness observed in both patients and mice suggests an acute (active, cellular) component rather than structural remodelling as potential cause of the loss of arterial compliance [9]. Interestingly, in the present study, we observed an initial increase in PWV in lenvatinib-treated mice on day 2 of treatment. However, when comparing ex vivo Peterson’s elastic modulus as index of arterial stiffness in response to lenvatinib after 4-day treatment in mice, no changes were detected. The pressure-stiffness relationship under DEANO, which reflects the contribution of matrix proteins to the stiffness of the aortic wall (i.e.; passive stiffness), was unaltered. Taken together, this data demonstrated that no changes occurred in structural stiffness, suggesting functional changes as a cause for arterial stiffening.
Importantly, the initial increase in PWV (day 2) was reversible after discontinuation of lenvatinib (day 8) and could be linked to the increase in peripheral blood pressure in our mice. This is in line with the hypothesis that increased PWV is primarily driven by functional changes rather than structural alterations in the arterial wall.
Vascular reactivity and endothelial function
Endothelium-independent relaxation in response to DEANO was unaltered in the lenvatinib-treated group, indicating a preserved reactivity of VSMCs to NO and intact VSMC function. Several studies associate the development of TKI-induced hypertension with reduced NO bioavailability and endothelial dysfunction [11, 14, 33]. In particular, reduced urinary excretion of NO metabolites was reported in rats treated with sunitinib [11]. Additionally, vatalanib showed decreased NO production and impaired eNOS phosphorylation in human aortic ECs [33]. Although some investigations support the hypothesis that vascular complications in response to TKIs are mediated by impaired endothelial function and NO deficiency [11, 14, 33], our ex vivo findings do not confirm this.
Our findings in the thoracic descending aorta align with previous studies where sunitinib treatment did not show any alteration of ACh-mediated vasodilation in iliac artery segments of rats [34, 35]. Despite vatalanib-receiving patients showing increased levels of plasma NO metabolites, flow-mediated dilation as index of NO bioavailability remained unchanged [36]. Altogether, the current conflicting evidence makes it difficult to conclude whether TKI-induced hypertension primarily arises from impaired NO signalling. The dissimilarities among these studies may be due to differences in the models used (C57Bl/6J mice vs male Wistar Kyoto rats [11, 34] vs human aortic ECs [33]) or in the treatment dose and duration. The present study used a 4-day treatment with lenvatinib (4 mg/kg/day) as an intravenous injection, while others used an 8-day treatment with sunitinib (14 mg/kg/day) [34], an 8-day treatment with sunitinib (26.7 mg/kg/day) as oral gavage [11], human aortic ECs stimulated with vatalanib (100 nmol/L) or mice treated for 2 weeks with vatalanib (100 mg/kg/day) [33]. Additionally, TKIs have different relative CV risk profiles when used in human patients [37,38,39,40].
ATP-induced vasorelaxation
Lenvatinib increased ATP-mediated relaxation, which is acting through P2Yx and P2X receptors [41]. Interestingly, Gast et al. showed that ATP and VEGF165a, an angiogenic factor, act synergistically on ECs, and the resulting complex determines an increased interaction with P2Yx receptors [42, 43], suggesting P2Yx receptors are recruited by TKIs targeting VEGF-signalling. Although the exact nature of this conformational change is still unknown [42], it may explain the augmented ATP-induced relaxation observed in our study. The enhanced ATP-mediated relaxation could indicate compensatory mechanisms. In agreement with this hypothesis, P2Y2 and P2Y6 receptors were upregulated. Hypertension may reduce shear stress, which in turn decreases ATP release [44]. To compensate for the reduced ATP availability, the aorta may upregulate P2Y receptors [45], improving relaxation.
Indeed, increased VEGF165a levels, due to the VEGFR-2 inhibition by lenvatinib, forming a synergistic complex with ATP, would cause a stronger response to the nucleotide via P2Yx receptor signalling. Moreover, the increased mRNA expression of both P2Y2 and P2Y6 receptors in the aortic segments of lenvatinib-treated mice suggests a compensatory role of purinergic signalling in the vascular responses associated with this TKI. Additionally, we have shown that treatment interruption leads to a recovery in ATP-dependent relaxation and normalisation of P2Yx receptor expression.
Cardiac alterations
Left ventricular dysfunction and cardiomyocyte hypertrophy have been reported in patients receiving TKIs targeting VEGFR-2 [38, 46]. Using echocardiography, this study aimed to comprehensively characterise the effect of lenvatinib on cardiac function in mice. TKIs have been associated with hypertension [47], but they differ in terms of their impact on the heart [38]. Echocardiographic evaluation in the present study revealed a marked reduction of left ventricular systolic, but not diastolic function in response to lenvatinib, which was reversible after treatment interruption. In addition to evaluating systolic and diastolic performances, a significant enlargement of the left ventricle was observed in lenvatinib-treated mice, along with a considerable reduction of systolic ventricular wall thickness. Echocardiographic features are suggestive of lenvatinib-induced dilated cardiomyopathy [48]. Clinical data about lenvatinib-associated cardiomyopathy is rather limited. In this context, Takotsubo cardiomyopathy with severe left ventricular dysfunction has been reported in a patient receiving lenvatinib for thyroid cancer [49]. Lenvatinib has also been associated with non-ischemic cardiomyopathy in a case report [50]. To investigate whether the echocardiographic alterations were correlated with changes at a cellular level, histology was performed to quantify cardiomyocyte size. A larger cardiomyocyte cross-sectional area observed in lenvatinib-treated animals supports the presence of cardiac dilation/(or) hypertrophy. The cardiomyocyte enlargement in response to lenvatinib can be hypothesised to be an adaptive response to compensate for the reduced LVEF by increasing wall thickness and preserving systolic function, according to Laplace relationship [51]. The resultant hypertrophic remodelling, characterised by an initial reactive increase in wall thickness, may become maladaptive [51]. Interestingly, in swine, a similar increase in afterload and vascular resistance, accompanied by early hypertrophic remodelling was demonstrated within one week of sunitinib therapy [52]. The induction of hypertension and increase in afterload prompt the heart to undergo hypertrophic remodelling, which elevates oxygen demand. Further triggering the activation of the HIF-1/α-VEGF axis, which is typically recruited to meet the heightened metabolic needs by promoting angiogenesis [52, 53]. VEGF inhibition, however, may disrupt this balance, as seen in TKI-treated models and patients, potentially exacerbating myocardial ischemia and dysfunction [54,55,56].
However, attributing such a response (after 4 days of treatment) to remodelling is ambiguous, as other acute cellular responses such as myocyte swelling could eventually lead to similar observations.
Limitations
This study primarily focused on the short-term effects of lenvatinib and the interplay between vascular stiffness and cardiac function. While the acute and brief treatment regimen provides valuable insights into early hemodynamic changes, it does not replicate the chronic exposure experienced by patients undergoing long-term therapy. Longer treatment protocols would be crucial to understand the full spectrum of lenvatinib's toxicity and potential reversibility. Additionally, future studies should prioritise investigating renal function alongside cardiovascular outcomes to provide a more comprehensive understanding of lenvatinib's side effects.
Conclusion
Lenvatinib induced hypertension and reduced LVEF in vivo. These findings underscore the importance of monitoring CV health in patients undergoing TKI therapy, particularly those receiving lenvatinib. Additionally, the rapid onset of hypertension and arterial stiffness in lenvatinib-treated mice indicates an acute vascular response although its mechanism remains elusive. Interestingly, arterial stiffness was reversible once treatment was ceased. Furthermore, lenvatinib-enhanced ATP-mediated relaxation, further proposing a potential involvement of P2Yx receptors in the vascular response to the TKI, which was supported by an increase in receptor expression. Importantly, cessation of treatment led to recovery in ATP-dependent relaxation and normalisation of P2Yx receptor expression, suggesting a reversible effect on purinergic signalling pathways.
Data availability
No datasets were generated or analysed during the current study.
Abbreviations
- ACh:
-
Acetylcholine
- ATP:
-
Adenosine triphosphate
- CV:
-
Cardiovascular
- DEANO:
-
Diethylamine NONOate
- EC:
-
Endothelial cells
- eNOS:
-
Endothelial nitric oxide synthase
- L-NAME:
-
Nω-nitro-L-arginine methyl ester
- LVEF:
-
Left ventricular ejection fraction
- LVP:
-
Left ventricular pressure
- NO:
-
Nitric oxide
- ns:
-
Not significant
- PE:
-
Phenylephrine
- PWV:
-
Pulse wave velocity
- qRT-PCR:
-
Quantitative real-time polymerase chain reaction
- ROTSAC:
-
Rodent Oscillatory Set-up to Study Arterial Compliance
- TKI:
-
Tyrosine kinase inhibitor
- VEGF:
-
Vascular endothelial growth factor
- VEGFR-2:
-
Vascular endothelial growth factor receptor-2
- VSMCs:
-
Vascular smooth muscle cells
References
Shibuya M. Vascular Endothelial Growth Factor (VEGF) and Its Receptor (VEGFR) Signaling in Angiogenesis: A Crucial Target for Anti- and Pro-Angiogenic Therapies. Genes Cancer. 2011;2:1097–105.
Ferrara N. Adamis AP (2016) Ten years of anti-vascular endothelial growth factor therapy. Nat Rev Drug Discov. 2016;156(15):385–403.
Ferrara N. Vascular endothelial growth factor: Basic science and clinical progress. Endocr Rev. 2004;25:581–611.
Lyon AR, López-Fernánde T, Couch LS, et al. 2022 ESC Guidelines on cardio-oncology developed in collaboration with the European Hematology Association (EHA), the European Society for Therapeutic Radiology and Oncology (ESTRO) and the International Cardio-Oncology Society (IC-OS). Eur Heart J. 2022;43:4229–361.
Hussein Z, Mizuo H, Hayato S, Namiki M, Shumaker R. Clinical Pharmacokinetic and Pharmacodynamic Profile of Lenvatinib, an Orally Active, Small-Molecule, Multitargeted Tyrosine Kinase Inhibitor. Eur J Drug Metab Pharmacokinet. 2017;42:903–14.
Hu-Lowe DD, Zou HY, Grazzini ML, et al. Nonclinical Antiangiogenesis and Antitumor Activities of Axitinib (AG-013736), an Oral, Potent, and Selective Inhibitor of Vascular Endothelial Growth Factor Receptor Tyrosine Kinases 1, 2, 3. Clin Cancer Res. 2008;14:7272–83.
Rehman O, Jaferi U, Padda I, Khehra N, Atwal H, Mossabeh D, Bhangu R. Overview of lenvatinib as a targeted therapy for advanced hepatocellular carcinoma. Clin Exp Hepatol. 2021;7:249–57.
Lankhorst S, Kappers MHW, Van Esch JHM, Danser AHJ, Van Den Meiracker AH. Hypertension during vascular endothelial growth factor inhibition: Focus on nitric oxide, endothelin-1, and oxidative stress. Antioxidants Redox Signal. 2014;20:135–45.
Camarda N, Travers R, Yang VK, London C, Jaffe IZ. VEGF Receptor Inhibitor-Induced Hypertension: Emerging Mechanisms and Clinical Implications. Curr Oncol Rep. 2022;24:463–74.
Steeghs N, Gelderblom H, Roodt JOT, Christensen O, Rajagopalan P, Hovens M, Putter H, Rabelink TJ, De Koning E. Hypertension and Rarefaction during Treatment with Telatinib, a Small Molecule Angiogenesis Inhibitor. Clin Cancer Res. 2008;14:3470–6.
Kappers MHW, Smedts FMM, Horn T, Van Esch JHM, Sleijfer S, Leijten F, Wesseling S, Strevens H, Danser AHJ, Van Den Meiracker AH. The vascular endothelial growth factor receptor inhibitor sunitinib causes a preeclampsia-like syndrome with activation of the endothelin system. Hypertension. 2011;58:295–302.
Mayer EL, Dallabrida SM, Rupnick MA, Redline WM, Hannagan K, Ismail NS, Burstein HJ, Beckman JA. Contrary effects of the receptor tyrosine kinase inhibitor vandetanib on constitutive and flow-stimulated nitric oxide elaboration in humans. Hypertension. 2011;58:85–92.
Lankhorst S, Saleh L, Danser AHJ, Van Den Meiracker AH. Etiology of angiogenesis inhibition-related hypertension. Curr Opin Pharmacol. 2015;21:7–13.
Eechoute K, Van Der Veldt AAM, Oosting S, et al. Polymorphisms in endothelial nitric oxide synthase (eNOS) and vascular endothelial growth factor (VEGF) predict sunitinib-induced hypertension. Clin Pharmacol Ther. 2012;92:503–10.
Pannucci P, Van Daele M, Cooper SL, Wragg ES, March J, Groenen M, Hill SJ, Woolard J. Role of endothelin ETA receptors in the hypertension induced by the VEGFR-2 kinase inhibitors axitinib and lenvatinib in conscious freely-moving rats. Biochem Pharmacol. 2024;228:116007.
Veronese ML, Mosenkis A, Flaherty KT, Gallagher M, Stevenson JP, Townsend RR, O’Dwyer PJ. Mechanisms of Hypertension Associated With BAY 43–9006. J Clin Oncol. 2006;24:1363–9.
Catino AB, Hubbard RA, Chirinos JA, et al. Longitudinal Assessment of Vascular Function With Sunitinib in Patients With Metastatic Renal Cell Carcinoma. Circ Heart Fail. 2018;11:e004408.
Mihalcea D, Memis H, Mihaila S, Vinereanu D. Cardiovascular Toxicity Induced by Vascular Endothelial Growth Factor Inhibitors. Life. 2023;366(13):366.
Maurea N, Coppola C, Piscopo G, Galletta F, Riccio G, Esposito E, De Lorenzo C, De Laurentiis M, Spallarossa P, Mercuro G. Pathophysiology of cardiotoxicity from target therapy and angiogenesis inhibitors. J Cardiovasc Med. 2016;17:S19–26.
Dobbin SJH, Cameron AC, Petrie MC, Jones RJ, Touyz RM, Lang NN. Toxicity of cancer therapy: what the cardiologist needs to know about angiogenesis inhibitors. Heart. 2018;104:1995–2002.
Jin Y, Xu Z, Yan H, He Q, Yang X, Luo P. A Comprehensive Review of Clinical Cardiotoxicity Incidence of FDA-Approved Small-Molecule Kinase Inhibitors. Front Pharmacol. 2020;11:891.
Krüger DN, Bosman M, Van Assche CXL, et al. Characterization of systolic and diastolic function, alongside proteomic profiling, in doxorubicin-induced cardiovascular toxicity in mice. Cardio-Oncology. 2024;10:1–17.
Leloup AJA, Van Hove CE, Kurdi A, De Moudt S, Martinet W, De Meyer GRY, Schrijvers DM, De Keulenaer GW, Fransen P. A novel set-up for the ex vivo analysis of mechanical properties of mouse aortic segments stretched at physiological pressure and frequency. J Physiol. 2016;594:6105–15.
Carter JJ, Fretwell LV, Woolard J. Effects of 4 multitargeted receptor tyrosine kinase inhibitors on regional hemodynamics in conscious, freely moving rats. FASEB J. 2017;31:1193–203.
Ptinopoulou AG, Sprangers B. Tyrosine kinase inhibitor-induced hypertension—marker of anti-tumour treatment efficacy or cardiovascular risk factor? Clin Kidney J. 2021;14:14–7.
Ollero M, Sahali D. Inhibition of the VEGF signalling pathway and glomerular disorders. Nephrol Dial Transplant. 2015;30:1449–55.
Safar ME, Levy BI, Struijker-Boudier H. Current perspectives on arterial stiffness and pulse pressure in hypertension and cardiovascular diseases. Circulation. 2003;107:2864–9.
Laurent S, Boutouyrie P, Asmar R, Gautier I, Laloux B, Guize L, Ducimetiere P, Benetos A. Aortic stiffness is an independent predictor of all-cause and cardiovascular mortality in hypertensive patients. Hypertension. 2001;37:1236–41.
Kaess BM, Rong J, Larson MG, Hamburg NM, Vita JA, Levy D, Benjamin EJ, Vasan RS, Mitchell GF. Aortic Stiffness, Blood Pressure Progression, and Incident Hypertension. JAMA. 2012;308:875–81.
Van Varik BJ, Rennenberg RJMW, Reutelingsperger CP, Kroon AA, De Leeuw PW, Schurgers LJ. Mechanisms of arterial remodeling: Lessons from genetic diseases. Front Genet. 2012;3:290.
De Moudt S, Leloup A, Fransen P. Aortic Stiffness Hysteresis in Isolated Mouse Aortic Segments Is Intensified by Contractile Stimuli, Attenuated by Age, and Reversed by Elastin Degradation. Front Physiol. 2021;12:1–16.
Dumor K, Shoemaker-Moyle M, Nistala R, Whaley-Connell A. Arterial Stiffness in Hypertension: an Update. Curr Hypertens Rep. 2018;20:72.
Neves KB, Rios FJ, Van Der Mey L, Alves-Lopes R, Cameron AC, Volpe M, Montezano AC, Savoia C, Touyz RM. VEGFR (vascular endothelial growth factor receptor) inhibition induces cardiovascular damage via redox-sensitive processes. Hypertension. 2018;71:638–47.
Colafella KMM, Neves KB, Montezano AC, et al. Selective ETA vs. dual ETA/B receptor blockade for the prevention of sunitinib-induced hypertension and albuminuria in WKY rats. Cardiovasc Res. 2020;116:1779–90.
Mirabito Colafella KM, Van Dorst DCH, Neuman RI, et al. Differential effects of cyclo-oxygenase 1 and 2 inhibition on angiogenesis inhibitor-induced hypertension and kidney damage. Clin Sci. 2022;136:675–94.
Neishi Y, Mochizuki S, Miyasaka T, et al. Evaluation of bioavailability of nitric oxide in coronary circulation by direct measurement of plasma nitric oxide concentration. Proc Natl Acad Sci U S A. 2005;102:11456–61.
Motzer RJ, Taylor MH, Evans TRJ, et al. Lenvatinib dose, efficacy, and safety in the treatment of multiple malignancies. Expert Rev Anticancer Ther. 2022;22:383–400.
Shah RR, Morganroth J. Update on Cardiovascular Safety of Tyrosine Kinase Inhibitors: With a Special Focus on QT Interval, Left Ventricular Dysfunction and Overall Risk/Benefit. Drug Saf. 2015;38:693–710.
Rini BI, Quinn DI, Baum M, et al. Hypertension among patients with renal cell carcinoma receiving axitinib or sorafenib: analysis from the randomized phase III AXIS trial. Target Oncol. 2015;10:45–53.
Abdel-Rahman O, Fouad M. Risk of Cardiovascular Toxicities in Patients with Solid Tumors Treated with Sorafenib: An Updated Systematic Review and Meta-Analysis. Futur Oncol. 2014;10:1981–92.
Ralevic V, Burnstock G. Receptors for purines and pyrimidines. Pharmacol Rev. 1998;50:413–92.
Gast RE, König S, Rose K, Ferenz KB, Krieglstein J. Binding of ATP to vascular endothelial growth factor isoform VEGF-A165 is essential for inducing proliferation of human umbilical vein endothelial cells. BMC Biochem. 2011;12:28.
Buxton ILO, Rumjahn SM, Yokdang N, Baldwin KA, Thai J. Purinergic regulation of vascular endothelial growth factor signaling in angiogenesis. Br J Cancer. 2009;100:1465–70.
Yamamoto K, Sokabe T, Matsumoto T, et al. (2005) Impaired flow-dependent control of vascular tone and remodeling in P2X4-deficient mice. Nat Med. 2005;121(12):133–7.
Wang L, Karlsson L, Moses S, Hultgårdh-Nilsson A, Andersson M, Borna C, Gudbjartsson T, Jern S, Erlinge D. P2 receptor expression profiles in human vascular smooth muscle and endothelial cells. J Cardiovasc Pharmacol. 2002;40:841–53.
Chu TF, Rupnick MA, Kerkela R, et al. Cardiotoxicity Associated with the Tyrosine Kinase Inhibitor Sunitinib. Lancet (London, England). 2007;370:2011.
Møller NB, Budolfsen C, Grimm D, Krüger M, Infanger M, Wehland M, Magnusson NE. Drug-Induced Hypertension Caused by Multikinase Inhibitors (Sorafenib, Sunitinib, Lenvatinib and Axitinib) in Renal Cell Carcinoma Treatment. Int J Mol Sci. 2019;4712(20):4712.
Faggiano A, Avallone C, Gentile D, Provenzale G, Toriello F, Merlo M, Sinagra G, Carugo S. Echocardiographic advances in dilated cardiomyopathy. J Clin Med. 2021;10:5518.
Chae YK, Chiec L, Adney SK, Waitzman J, Costa R, Carneiro B, Matsangou M, Agulnik M, Kopp P, Giles F. Posterior reversible encephalopathy syndrome and takotsubo cardiomyopathy associated with lenvatinib therapy for thyroid cancer: a case report and review. Oncotarget. 2018;9:28281–9.
Matsuo M, Wakasaki T, Yasumatsu R, Nakagawa T. A case of cardiotoxicity developed under the administration of lenvatinib in a thyroid cancer patient. Pract Otorhinolaryngol (Basel). 2020;113:309–14.
Diwan A, Dorn GW. Decompensation of cardiac hypertrophy: Cellular mechanisms and novel therapeutic targets. Physiology. 2007;22:56–64.
Kappers MHW, De Beer VJ, Zhou Z, Danser AHJ, Sleijfer S, Duncker DJ, Van Den Meiracker AH, Merkus D. Sunitinib-induced systemic vasoconstriction in swine is endothelin mediated and does not involve nitric oxide or oxidative stress. Hypertension. 2012;59:151–7.
Oka T, Akazawa H, Naito AT, Komuro I. Angiogenesis and cardiac hypertrophy: Maintenance of cardiac function and causative roles in heart failure. Circ Res. 2014;114:565–71.
Steeghs N, Rabelink TJ, Op ’t Roodt J, Batman E, Cluitmans FHM, Weijl NI, de Koning E, Gelderblom H,. Reversibility of capillary density after discontinuation of bevacizumab treatment. Ann Oncol Off J Eur Soc Med Oncol. 2010;21:1100–5.
Chintalgattu V, Rees ML, Culver JC, et al. Coronary microvascular pericytes are the cellular target of sunitinib malate-induced cardiotoxicity. Sci Transl Med. 2013. https://doiorg.publicaciones.saludcastillayleon.es/10.1126/SCITRANSLMED.3005066.
Izumiya Y, Shiojima I, Sato K, Sawyer DB, Colucci WS, Walsh K. Vascular Endothelial Growth Factor Blockade Promotes the Transition From Compensatory Cardiac Hypertrophy to Failure in Response to Pressure Overload. Hypertension. 2006;47:887–93. https://doiorg.publicaciones.saludcastillayleon.es/10.1161/01.HYP.0000215207.54689.31.
Acknowledgements
The Laboratory of Physiopharmacology and Department of Cardiology are part of the Infla-Med Centre of Excellence, University of Antwerp, Antwerp, Belgium.
Funding
This project has received funding from the European Union’s Horizon 2020 research and innovation programme under the Marie Skłodowska-Curie grant agreement No 858070 and the Belgian Foundation against Cancer (research grant C/2020/1374) C.F. is holder of a clinical mandate from the Belgian Foundation against Cancer (grant number: 2021–034) and was awarded a research grant from the King Baudouin Foundation (Funds Pierre Masure, Alphonse & Marie Walckiers, and De Winter-Vermant 2018). The Vevo LAZR-X device was acquired thanks to the National Fund for Scientific Research – Flanders (FWO), grant I005122N. Furthermore, the study is supported by a DOCPRO4 grant (BOF UAntwerp ID: 39984). D.N.K. received funding from the University of Antwerp (BOF UAntwerp ID: 49195). C.D.W received funding from the University of Antwerp (BOF UAntwerp ID: 49194).
Author information
Authors and Affiliations
Contributions
Conceptualisation: P.P., D.N.K., G.R.Y.D.M., W.M., P.J.G, S.J.H., C.F., and J.W. Methodology: D.N.K., P.P., C.H.G.N., and C.D.W Investigation and formal analysis: P.P, D.N.K., C.H.G.N., C.D.W., and P.J.G. Writing-original draft preparation: P.P., D.N.K, J.W. S.J.H., C.F and P.J.G Funding acquisition: G.R.Y.D.M., W.M., J.W., S.J.H., C.F., and P.J.G. All authors have read and reviewed the manuscript and agreed to the published version of the manuscript.
.
Corresponding author
Ethics declarations
Ethics approval and consent to participate
Use of animals approval of the Ethical Committee for Animal Testing of the University of Antwerp (ethical file 2022–40) conformed with the ARRIVE guidelines, under Directive 2010/63/EU, and with the Belgian Royal Decree of 2013 and the Guide for the Care and Use of Laboratory Animals published by the US National Institutes of Health (NIH Publication no.85–23, revised 1996).
Welfare of animals was assessed daily by the animal caretakers and the principal researcher based on the Functional Observation Battery scoring system. Criteria for humane euthanasia were as follows: Animals displaying signs of pain based on the scoring table, meeting specific score thresholds, or exhibiting significant weight loss (> 20%).
Consent for publication
All authors have read and reviewed the manuscript and agreed to the published version of the manuscript.
Competing interests
The authors declare no competing interests.
Additional information
Publisher’s Note
Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Supplementary Information
Rights and permissions
Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.
About this article
Cite this article
Krüger, D.N., Pannucci, P., Wesley, C.D. et al. Acute vascular and cardiac effects of lenvatinib in mice. Cardio-Oncology 11, 14 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s40959-025-00307-8
Received:
Accepted:
Published:
DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s40959-025-00307-8